Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 609
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Inorg Chem ; 63(23): 10455-10465, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38743433

RESUMO

Organomercurials (RHg+), especially methylmercury (MeHg+) and ethylmercury (EtHg+), are considered to be more neurotoxic than the inorganic counterpart (Hg2+). They cause massive DNA damage in cells, especially in neurons, where cellular glutathione (GSH) levels are significantly low. However, the mechanism by which RHg+ exerts massive DNA damage at cytotoxic concentrations in brain cells remains obscure. In this study, we investigated the effect of RHg+ on the structural and electronic properties of nucleosides and its effects on DNA damage. The direct interaction of RHg+ with the nucleoside significantly weakens N-glycosidic bonds, decreases the C-H bond energy of sugar moieties, and increases the electrophilicity of the C8-center of purine bases. As a consequence, RHg+-conjugated DNA molecules are extremely labile and highly sensitive to any nucleophiles/radicals present in GSH-depleted cells and, thus, undergo enhanced oxidative and unusual alkylative DNA damage. We also report a functional model of organomercurial lyase, which showed excellent cytoprotective effect against RHg+-induced cytotoxicity; this reverses the activity of glutathione reductase inhibited by MeHgCl and ceases oxidative and alkylating DNA damage. This intriguing finding provides new mechanistic insight into the mode of action of organomercurials in GSH-depleted cells and their adverse effects on individuals with neurodegenerative disorders associated with oxidative stress.


Assuntos
Dano ao DNA , Glutationa , Compostos de Metilmercúrio , Compostos de Metilmercúrio/farmacologia , Compostos de Metilmercúrio/química , Glutationa/metabolismo , Glutationa/química , Dano ao DNA/efeitos dos fármacos , Humanos , DNA/química , DNA/efeitos dos fármacos , Estrutura Molecular , Animais , Sobrevivência Celular/efeitos dos fármacos , Teoria da Densidade Funcional
2.
Chem Res Toxicol ; 35(1): 77-88, 2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-34905692

RESUMO

Metabolic effects of methylmercury (MeHg) are gaining wider attention. We have previously shown that MeHg causes lipid dysregulation in Caenorhabditis elegans (C. elegans), leading to altered gene expression, increased triglyceride levels and lipid storage, and altered feeding behaviors. Transcriptional regulators, such as transcription factors and microRNAs (miRNAs), have been shown to regulate lipid storage, serum triglycerides, and adipogenic gene expression in human and rodent models of metabolic diseases. As we recently investigated adipogenic transcription factors induced by MeHg, we were, therefore, interested in whether MeHg may also regulate miRNA sequences to cause metabolic dysfunction. Lipid dysregulation, as measured by triglyceride levels, lipid storage sites, and feeding behaviors, was assessed in wild-type (N2) worms and in transgenic worms that either were sensitive to miRNA expression or were unable to process miRNAs. Worms that were sensitive to the miRNA expression were protected from MeHg-induced lipid dysregulation. In contrast, the mutant worms that were unable to process miRNAs had exacerbated MeHg-induced lipid dysregulation. Concurrent with differential lipid homeostasis, miRNA-expression mutants had altered MeHg-induced mitochondrial toxicity as compared to N2, with the miRNA-sensitive mutants showing mitochondrial protection and the miRNA-processing mutants showing increased mitotoxicity. Taken together, our data demonstrate that the expression of miRNAs is an important determinant in MeHg toxicity and MeHg-induced metabolic dysfunction in C. elegans.


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Compostos de Metilmercúrio/farmacologia , MicroRNAs/genética , Mitocôndrias/efeitos dos fármacos , Animais , Caenorhabditis elegans/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/genética , Metabolismo dos Lipídeos , Compostos de Metilmercúrio/química , Mitocôndrias/metabolismo , Relação Estrutura-Atividade
3.
Int J Mol Sci ; 23(5)2022 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-35270009

RESUMO

Methylmercury (MeHg), a long-lasting organic pollutant, is known to induce cytotoxic effects in mammalian cells. Epidemiological studies have suggested that environmental exposure to MeHg is linked to the development of diabetes mellitus (DM). The exact molecular mechanism of MeHg-induced pancreatic ß-cell cytotoxicity is still unclear. Here, we found that MeHg (1-4 µM) significantly decreased insulin secretion and cell viability in pancreatic ß-cell-derived RIN-m5F cells. A concomitant elevation of mitochondrial-dependent apoptotic events was observed, including decreased mitochondrial membrane potential and increased proapoptotic (Bax, Bak, p53)/antiapoptotic (Bcl-2) mRNA ratio, cytochrome c release, annexin V-Cy3 binding, caspase-3 activity, and caspase-3/-7/-9 activation. Exposure of RIN-m5F cells to MeHg (2 µM) also induced protein expression of endoplasmic reticulum (ER) stress-related signaling molecules, including C/EBP homologous protein (CHOP), X-box binding protein (XBP-1), and caspase-12. Pretreatment with 4-phenylbutyric acid (4-PBA; an ER stress inhibitor) and specific siRNAs for CHOP and XBP-1 significantly inhibited their expression and caspase-3/-12 activation in MeHg-exposed RIN-mF cells. MeHg could also evoke c-Jun N-terminal kinase (JNK) activation and reactive oxygen species (ROS) generation. Antioxidant N-acetylcysteine (NAC; 1mM) or 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (trolox; 100 µM) markedly prevented MeH-induced ROS generation and decreased cell viability in RIN-m5F cells. Furthermore, pretreatment of cells with SP600125 (JNK inhibitor; 10 µM) or NAC (1 mM) or transfection with JNK-specific siRNA obviously attenuated the MeHg-induced JNK phosphorylation, CHOP and XBP-1 protein expression, apoptotic events, and insulin secretion dysfunction. NAC significantly inhibited MeHg-activated JNK signaling, but SP600125 could not effectively reduce MeHg-induced ROS generation. Collectively, these findings demonstrate that the induction of ROS-activated JNK signaling is a crucial mechanism underlying MeHg-induced mitochondria- and ER stress-dependent apoptosis, ultimately leading to ß-cell death.


Assuntos
Estresse do Retículo Endoplasmático , Compostos de Metilmercúrio , Animais , Apoptose , Caspase 3/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Mamíferos/metabolismo , Compostos de Metilmercúrio/farmacologia , Mitocôndrias/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
4.
Int J Mol Sci ; 22(4)2021 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-33567754

RESUMO

The placental barrier can protect the fetus from contact with harmful substances. The potent neurotoxin methylmercury (MeHg), however, is very efficiently transported across the placenta. Our previous data suggested that L-type amino acid transporter (LAT)1 is involved in placental MeHg uptake, accepting MeHg-L-cysteine conjugates as substrate due to structural similarity to methionine. The aim of the present study was to investigate the antioxidant defense of placental cells to MeHg exposure and the role of LAT1 in this response. When trophoblast-derived HTR-8/SVneo cells were LAT1 depleted by siRNA-mediated knockdown, they accumulated less MeHg. However, they were more susceptible to MeHg-induced toxicity. This was evidenced in decreased cell viability at a usually noncytotoxic concentration of 0.03 µM MeHg (~6 µg/L). Treatment with ≥0.3 µM MeHg increased cytotoxicity, apoptosis rate, and oxidative stress of HTR-8/SVneo cells. These effects were enhanced under LAT1 knockdown. Reduced cell number was seen when MeHg-exposed cells were cultured in medium low in cysteine, a constituent of the tripeptide glutathione (GSH). Because LAT1-deficient HTR-8/SVneo cells have lower GSH levels than control cells (independent of MeHg treatment), we conclude that LAT1 is essential for de novo synthesis of GSH, required to counteract oxidative stress. Genetic predisposition to decreased LAT1 function combined with MeHg exposure could increase the risk of placental damage.


Assuntos
Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Compostos de Metilmercúrio/análise , Compostos de Metilmercúrio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Placenta/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Apoptose , Sobrevivência Celular , Células Cultivadas , Feminino , Glutationa/metabolismo , Humanos , Placenta/metabolismo , Placenta/patologia , Gravidez , Substâncias Protetoras/análise
5.
Inorg Chem ; 59(5): 2711-2718, 2020 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-32049511

RESUMO

Selenoenzymes, containing a selenocysteine (Sec) residue, fulfill important roles in biology. The mammalian thioredoxin reductase selenoenzymes are key regulators of antioxidant defense and redox signaling and are inhibited by methylmercury species and by the gold-containing drug auranofin. It has been proposed that such inhibition is mediated by metal binding to Sec in the enzyme. However, direct structural observations of these classes of inhibitors binding to selenoenzymes have been few to date. Here we therefore have used extended X-ray absorption fine structure as a direct structural probe to investigate binding to the selenium site in recombinant rat thioredoxin reductase 1 (TrxR1). The results demonstrate for the first time the direct and complete binding of the metal atom of the inhibitors to the selenium atom in TrxR1 for both methylmercury and auranofin, indicating that TrxR1 inhibition indeed can be attributed to such direct metal-selenium binding.


Assuntos
Auranofina/química , Auranofina/farmacologia , Compostos de Metilmercúrio/química , Compostos de Metilmercúrio/farmacologia , Selenocisteína/química , Tiorredoxinas/antagonistas & inibidores , Tiorredoxinas/química , Animais , Sítios de Ligação/efeitos dos fármacos , Ratos , Selenocisteína/metabolismo , Tiorredoxinas/metabolismo
6.
Int J Mol Sci ; 21(5)2020 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-32106432

RESUMO

Exposure to environmental endocrine disruptors has been associated with an increased frequency of thyroid pathology. In this study, we evaluated the effects of various concentrations of methylmercury (MeHg) on immortalized, non-tumorigenic thyroid cells (Nthy-ori-3-1). Exposure to MeHg at 2.5 and 5 µM for 24 h caused a reduction in cell viability with a decrease of the cell population in sub-G0 phase, as detected by MTT and flow cytometry. Conversely, MeHg at the lower concentration of 0.1 µM increased the cell viability with a rise of G2/M phase. An immunoblot analysis showed higher expression levels of phospho-ERK and not of phospho-Akt. Further enhancement of the cell growth rate was observed after a prolonged exposure of the cells up to 18 days to MeHg 0.1 µM. The present findings demonstrate the toxicity of high concentrations of MeHg on thyroid cells, while showing that treatment with lower doses of Hg, as may occur after prolonged exposure to this environmental contaminant, exerts a promoting effect on thyroid cell proliferation, by acting on the ERK-mediated pro-oncogenic signal transduction pathway.


Assuntos
Proliferação de Células , Disruptores Endócrinos/farmacologia , Sistema de Sinalização das MAP Quinases , Compostos de Metilmercúrio/farmacologia , Células Epiteliais da Tireoide/efeitos dos fármacos , Linhagem Celular , Humanos , Células Epiteliais da Tireoide/metabolismo , Células Epiteliais da Tireoide/fisiologia
7.
Toxicol Appl Pharmacol ; 362: 59-66, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30352208

RESUMO

Methylmercury (MeHg+) is an extremely toxic organomercury cation that can induce severe neurological damage. Once it enters the body, methylmercury binds to amino acids or proteins containing free sulfhydryl groups. In particular, methylmercury is known to bind with human serum albumin (HSA) in human plasma; however, the effects of methylmercury-HSA conjugate (MeHg-HSA) on the central nervous system (CNS) are not fully understood. In the present study, we used the microglial cell line N9 as the target cells to evaluate the effect of MeHg-HSA on physiological function of the CNS preliminarily. The various factors in the cell culture were monitored by MTT assay, total lactate dehydrogenase assay, ELISA, qPCR, Western blot and flow cytometry techniques. The results showed that low-dose treatment with MeHg-HSA activated N9 cells, promoting cell proliferation and total cell number, enhancing NO and intracellular Ca2+ levels, and suppressing the release of TNFα and IL1ß without cytotoxic effects; while high-dose MeHg-HSA exhibited cytotoxic effects on N9 cells, including promoting cell death and increasing the secretion of TNFα and IL1ß. These results indicate that MeHg-HSA causes hormesis in microglia N9 cells. Furthermore, ERK/MAPKs and STAT3 signaling pathways related to the hormesis of MeHg-HSA on N9 cells. In addition, low dose of MeHg-HSA might be viewed as something very close to a lowest observed adverse effect level (LOAEL) for N9 cells. These findings will be useful for investigating the hormesis mechanism of MeHg+ and exploring the specific functions of MeHg-sulfhydryl conjugates on the central nervous system.


Assuntos
Compostos de Metilmercúrio/farmacologia , Microglia/efeitos dos fármacos , Albumina Sérica Humana/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Hormese/efeitos dos fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Compostos de Metilmercúrio/química , Camundongos , Microglia/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico/metabolismo , Fator de Transcrição STAT3/metabolismo , Albumina Sérica Humana/química , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
8.
Chem Res Toxicol ; 32(8): 1656-1669, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31340646

RESUMO

Methylmercury (MeHg) and perfluorooctanesulfonate (PFOS) are major contaminants of human blood that are both common in dietary fish, thereby raising questions about their combined impact on human development. Here, pregnant Sprague-Dawley rats ingested a daily dose, from gestational day 1 through to weaning, of either 1 mg/kg bw PFOS (PFOS-only), 1 mg/kg MeHg (MeHg-only), a mixture of 0.1 mg/kg PFOS and 1 mg/kg MeHg (Low-Mix), or of 1 mg/kg of PFOS and 1 mg/kg MeHg (High-Mix). Newborns were monitored for physical milestones and reflexive developmental responses, and in juveniles the spontaneous activity, anxiety, memory, and cognition were assessed. Targeted metabolomics of 199 analytes was applied to sectioned brain regions of juvenile offspring. Newborns in the High-Mix group had decreased weight gain as well as delayed reflexes and innate behavioral responses compared to controls and individual chemical groups indicating a toxicological interaction on early development. In juveniles, cumulative mixture effects increased in a dose-dependent manner in tests of anxiety-like behavior. However, other developmental test results suggested antagonism, as PFOS-only and MeHg-only juveniles had increased hyperactivity and thigmotaxic behavior, respectively, but fewer effects in Low-Mix and High-Mix groups. Consistent with these behavioral observations, a pattern of antagonism was also observed in neurochemicals measured in rat cortex, as PFOS-only and MeHg-only juveniles had altered concentrations of metabolites (e.g., lipids, amino acids, and biogenic amines), while no changes were evident in the combined exposures. The cortical metabolites altered in PFOS-only and MeHg-only exposed groups are involved in inhibitory and excitatory neurotransmission. These proof-of-principle findings at relatively high doses indicate the potential for toxicological interaction between PFOS and MeHg, with developmental-stage specific effects. Future mixture studies at lower doses are warranted, and prospective human birth cohorts should consider possible confounding effects from PFOS and mercury exposure on neurodevelopment.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Fluorocarbonos/farmacologia , Metabolômica , Compostos de Metilmercúrio/farmacologia , Ácidos Alcanossulfônicos/administração & dosagem , Ácidos Alcanossulfônicos/análise , Animais , Encéfalo/patologia , Relação Dose-Resposta a Droga , Feminino , Fluorocarbonos/administração & dosagem , Fluorocarbonos/análise , Masculino , Compostos de Metilmercúrio/administração & dosagem , Compostos de Metilmercúrio/análise , Gravidez , Ratos , Ratos Sprague-Dawley
9.
J Biol Regul Homeost Agents ; 32(1): 147-151, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29504379

RESUMO

Glioma is the most common primary tumor in the brain, accounting for about 40~50% of intracranial primary tumors. Most chemotherapeutic drugs have difficulty in penetrating the blood-brain barrier, and their clinical applications are greatly limited. We evaluated the effects of methylmercury-L-cysteine (MeHg-L-cys) and methylmercury chloride (MMC) on apoptosis of C6 glioma cells. L-type amino acid transporter (LAT1) was used to investigate the targeted transport function and cytotoxicity of MeHg- L-cys in glioma. MeHg-L-cys enhanced the ability of targeting glioma cells and reduced the adverse reactions to normal brain tissues. Therefore, it is significantly important to develop new anti-glioma drugs targeting the blood-brain barrier.


Assuntos
Sistema y+L de Transporte de Aminoácidos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Glioma , Compostos de Metilmercúrio/farmacologia , Proteínas de Neoplasias/metabolismo , Animais , Glioma/tratamento farmacológico , Glioma/metabolismo , Glioma/patologia , Compostos de Metilmercúrio/química , Ratos
10.
Environ Res ; 167: 15-20, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30005196

RESUMO

Fetuses and neonates are known to be highly susceptible to methylmercury (MeHg) toxicity, but little is known about the relative uptake of MeHg from blood to the developing brain. We measured time-course changes in mercury (Hg) concentrations in the brain of fetal, neonate, weanling, and adult rats after an injection of 0.08 µg (0.4 nmol) Hg/g MeHg. In the prenatal experiment, MeHg was subcutaneously injected to pregnant dams on embryonic days 17, 18, 18.5, 19, 19.5, or 20, and Hg concentrations in tissues were measured in both mothers and fetuses on embryonic day 21 (1 day before parturition). Brain Hg levels in fetuses peaked 2 days after injection and were approximately 1.5 times higher than in mothers. In the postnatal experiment, the same MeHg dose was injected subcutaneously to male rats on postnatal days 1 (neonates), 35 (weanlings), or 56 (adults). Mercury concentrations in tissues were measured 1, 2, 3, 4, 5, or 6 days after the injection. Brain Hg levels peaked most rapidly in neonates, and were approximately 1.5 times higher than levels in weanlings or adults. Throughout the examined period, peak Hg levels in the brain and the Hg brain/blood ratio 24 h after injection were highest in fetuses, followed by the levels in neonates, and decreased with life stage. These findings suggest that relatively higher brain MeHg uptake is an important factor in the vulnerability of fetuses and neonates to MeHg exposure.


Assuntos
Encéfalo , Exposição Materna , Mercúrio , Compostos de Metilmercúrio , Animais , Feminino , Feto , Masculino , Compostos de Metilmercúrio/farmacocinética , Compostos de Metilmercúrio/farmacologia , Parto , Gravidez , Ratos
11.
Biol Pharm Bull ; 40(9): 1595-1598, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28867746

RESUMO

Methylmercury (MeHg) results in cell death through endoplasmic reticulum (ER) stress. Previously, we reported that MeHg induces S-mercuration at cysteine 383 or 386 in protein disulfide isomerase (PDI), and this modification induces the loss of enzymatic activity. Because PDI is a key enzyme for the maturation of nascent protein harboring a disulfide bond, the disruption in PDI function by MeHg results in ER stress via the accumulation of misfolded proteins. However, the effects of MeHg on unfolded protein response (UPR) sensors and their signaling remain unclear. In the present study, we show that UPR is regulated by MeHg. We found that MeHg specifically attenuated inositol-requiring enzyme 1α (IRE1α)-x-box binding protein 1 (XBP1) branch, but not the protein kinase RNA-like endoplasmic reticulum kinase (PERK) and activating transcriptional factor 6 (ATF6) branches. Treatment with GSK2606414, a specific PERK inhibitor, significantly inhibited MeHg-induced cell death. These findings suggest that MeHg exquisitely regulates UPR signaling involved in cell death.


Assuntos
Compostos de Metilmercúrio/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Fator 6 Ativador da Transcrição/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Camundongos , Proteína Dissulfeto Redutase (Glutationa)/metabolismo , Proteína 1 de Ligação a X-Box/antagonistas & inibidores , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/metabolismo
12.
J Pharmacol Exp Ther ; 356(1): 2-12, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26514794

RESUMO

Methylmercury (MeHg) disrupts cerebellar function, especially during development. Cerebellar granule cells (CGC), which are particularly susceptible to MeHg by unknown mechanisms, migrate during this process. Transient changes in intracellular Ca(2+) (Ca(2+) i) are crucial to proper migration, and MeHg is well known to disrupt CGC Ca(2+) i regulation. Acutely prepared slices of neonatal rat cerebellum in conjunction with confocal microscopy and fluo4 epifluorescence were used to track changes induced by MeHg in CGC Ca(2+) i regulation in the external (EGL) and internal granule cell layers (IGL) as well as the molecular layer (ML). MeHg caused no cytotoxicity but did cause a time-dependent increase in fluo4 fluorescence that depended on the stage of CGC development. CGCs in the EGL were most susceptible to MeHg-induced increases in fluo4 fluorescence. MeHg increased fluorescence in CGC processes but only diffusely; Purkinje cells rarely fluoresced in these slices. Neither muscimol nor bicuculline alone altered baseline fluo4 fluorescence in any CGC layer, but each delayed the onset and reduced the magnitude of effect of MeHg on fluo4 fluorescence in the EGL and ML. In the IGL, both muscimol and bicuculline delayed the onset of MeHg-induced increases in fluo4 fluorescence but did not affect fluorescence magnitude. Thus, acute exposure to MeHg causes developmental stage-dependent increases in Ca(2+) i in CGCs. Effects are most prominent in CGCs during development or early stages of migration. GABAA receptors participate in an as yet unclear manner to MeHg-induced Ca(2+) i dysregulation of CGCs.


Assuntos
Movimento Celular/efeitos dos fármacos , Cerebelo/citologia , Cerebelo/metabolismo , Compostos de Metilmercúrio/farmacologia , Receptores de GABA-A/efeitos dos fármacos , Compostos de Anilina , Animais , Animais Recém-Nascidos , Bicuculina/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/metabolismo , Feminino , Corantes Fluorescentes , Agonistas GABAérgicos/farmacologia , Antagonistas GABAérgicos/farmacologia , Técnicas In Vitro , Masculino , Muscimol/farmacologia , Gravidez , Células de Purkinje/efeitos dos fármacos , Ratos , Xantenos
13.
Environ Sci Technol ; 50(9): 4808-16, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27023211

RESUMO

Methylmercury (MeHg) is an established neurotoxicant of concern to fish-eating organisms. While most studies have focused on the fish consumers, much less is known about the effects of MeHg on the fish themselves, especially following exposures to chronic and environmentally relevant scenarios. Here we evaluated the behavioral effects of developmental MeHg insult by exposing parental generations of zebrafish to an environmentally realistic MeHg dietary concentration (1 ppm) and two higher concentrations (3 and 10 ppm) throughout their whole life span. Upon reaching adulthood, their offspring were analyzed through a series of behavioral tests, including the visual-motor response (VMR) assay, analysis of spontaneous swimming and evaluation of foraging efficiency. The VMR assay identified decreased locomotor output in the 6 day postfertilization (dpf) offspring of fish exposed to 3 and 10 ppm MeHg. However, in a second test 7 dpf fish revealed an increase in locomotor activity in all MeHg exposures tested. Increases in locomotion continued to be observed until 16 dpf, which coincided with increased foraging efficiency. These results suggest an association between MeHg and hyperactivity, and imply that fish chronically exposed to MeHg in the wild may be vulnerable to predation.


Assuntos
Comportamento Animal/efeitos dos fármacos , Peixe-Zebra , Animais , Dieta , Compostos de Metilmercúrio/farmacologia , Natação
14.
Biometals ; 29(3): 543-50, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27138944

RESUMO

Organoseleno-compounds have been investigated for its beneficial effects against methylmercury toxicity. In this way, diphenyl diselenide (PhSe)2 was demonstrated to decrease Hg accumulation in mice, protect against MeHg-induced mitochondrial dysfunction, and protect against the overall toxicity of this metal. In the present study we aimed to investigate if co-treatment with (PhSe)2 and MeHg could decrease accumulation of Hg in liver slices of rats. Rat liver slices were co-treated with (PhSe)2 (0.5; 5 µM) and/or MeHg (25 µM) for 30 min at 37 °C and Se and Hg levels were measured by inductively coupled plasma mass spectrometry (ICP-MS) in the slices homogenate, P1 fraction, mitochondria and incubation medium. Co-treatment with (PhSe)2 and MeHg did not significantly alter Se levels in any of the samples when compared with compounds alone. In addition, co-treatment with (PhSe)2 and MeHg did not decrease Hg levels in any of the samples tested, although, co-incubation significantly increased Hg levels in homogenate. We suggest here that (PhSe)2 could exert its previously demonstrated protective effects not by reducing MeHg levels, but forming a complex with MeHg avoiding it to bind to critical molecules in cell.


Assuntos
Derivados de Benzeno/farmacologia , Fígado/química , Fígado/efeitos dos fármacos , Mercúrio/análise , Compostos de Metilmercúrio/farmacologia , Compostos Organosselênicos/farmacologia , Selênio/análise , Animais , Derivados de Benzeno/administração & dosagem , Masculino , Espectrometria de Massas , Compostos de Metilmercúrio/administração & dosagem , Mitocôndrias Hepáticas/química , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Compostos Organosselênicos/administração & dosagem , Ratos , Ratos Wistar
15.
Biol Pharm Bull ; 39(8): 1353-8, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27476942

RESUMO

Methylmercury (MeHg) is one of the most toxic environmental pollutants and presents a serious hazard to health worldwide. Although the adverse effects of MeHg, including neurotoxicity, have been studied, its effects on immune function, in particular the immune response, remain unclear. This study examined the effects of low-dose MeHg on immune responses in mice. Mice were orally immunized with ovalbumin (OVA) or subcutaneously injected with mite extract to induce a T-helper 2 (Th2) allergic response. They were then exposed to MeHg (0, 0.02, 1.0, or 5.0 mg·kg(-1)·d(-1)). Immunization with oral OVA or subcutaneous mite extract increased serum levels of OVA-specific immunoglobulin (Ig) E (OVA-IgE), OVA-IgG1, interleukin (IL)-4, and IL-13, and total IgE, total IgG, and IL-13 when compared with levels in non-immunized mice. However, no interferon (IFN)-γ was detected. By contrast, serum levels of OVA-IgE, OVA-IgG1, IL-4, and IL-13, or total IgE, total IgG, and IL-13 in Th2 allergy model mice subsequently treated with MeHg were no higher than those in MeHg-untreated mice. These results suggest that MeHg exposure has no adverse effects on Th2 immune responses in antigen-immunized mice.


Assuntos
Dermatophagoides pteronyssinus/imunologia , Hipersensibilidade/imunologia , Compostos de Metilmercúrio/farmacologia , Ovalbumina/imunologia , Células Th2/imunologia , Animais , Encéfalo/metabolismo , Citocinas/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Imunoglobulina E/sangue , Imunoglobulina E/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C
16.
Int J Mol Sci ; 17(12)2016 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-27941687

RESUMO

Mitochondria are essential organelles and important targets for environmental pollutants. The detection of mitochondrial biogenesis and generation of reactive oxygen species (ROS) and p53 levels following low-dose methylmercury (MeHg) exposure could expand our understanding of underlying mechanisms. Here, the sensitivity of immortalized human neural progenitor cells (ihNPCs) upon exposure to MeHg was investigated. We found that MeHg altered cell viability and the number of 5-ethynyl-2'-deoxyuridine (EdU)-positive cells. We also observed that low-dose MeHg exposure increased the mRNA expression of cell cycle regulators. We observed that MeHg induced ROS production in a dose-dependent manner. In addition, mRNA levels of peroxisome-proliferator-activated receptor gammacoactivator-1α (PGC-1α), mitochondrial transcription factor A (TFAM) and p53-controlled ribonucleotide reductase (p53R2) were significantly elevated, which were correlated with the increase of mitochondrial DNA (mtDNA) copy number at a concentration as low as 10 nM. Moreover, we examined the expression of microRNAs (miRNAs) known as regulatory miRNAs of p53 (i.e., miR-30d, miR-1285, miR-25). We found that the expression of these miRNAs was significantly downregulated upon MeHg treatment. Furthermore, the overexpression of miR-25 resulted in significantly reducted p53 protein levels and decreased mRNA expression of genes involved in mitochondrial biogenesis regulation. Taken together, these results demonstrated that MeHg could induce developmental neurotoxicity in ihNPCs through altering mitochondrial functions and the expression of miRNA.


Assuntos
Compostos de Metilmercúrio/farmacologia , MicroRNAs/genética , Mitocôndrias/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , DNA Mitocondrial/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Células-Tronco Neurais/citologia , Biogênese de Organelas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ribonucleotídeo Redutases/metabolismo , Fatores de Transcrição/metabolismo
17.
J Neurosci Res ; 92(1): 95-103, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24123177

RESUMO

Astrocytes are actively involved in brain development, in mature CNS regulation, and in brain plasticity. They play a critical role in response to cerebral injuries and toxicants through a reaction known as "reactive gliosis," which is characterized by specific structural and functional features. A large amount of literature highlights the central role of astrocytes in mediating methylmercury (MeHg) neurotoxicity. In fact, mercury is the major neurotoxic pollutant that continues to arouse interest in research because of the severe risk it poses to human health. In this article, we focus on the action of MeHg on human astrocyte (HA) reactivity. We clearly demonstrate that MeHg induces a state of cellular suffering by promoting delayed and atypical astrocyte reactivity mediated by impairment of the proliferative and trophic component of the astrocyte together with an inflammatory state. This condition is generated by negative modulation of the major proteins of the filamentous network, which is manifested by the destabilization of astrocytic cytoarchitecture. Our data confirms the toxic effects of MeHg on HA reactivity and allows us to hypothesize that the establishment of this state of suffering and the delayed onset of a typical astrocytic reactivity compromise the main protective function of HA.


Assuntos
Astrócitos/efeitos dos fármacos , Gliose/induzido quimicamente , Compostos de Metilmercúrio/farmacologia , Astrócitos/patologia , Linhagem Celular , Gliose/patologia , Humanos
18.
Chem Res Toxicol ; 27(2): 254-64, 2014 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-24397474

RESUMO

Methylmercury (CH3Hg) is one of the forms of mercury found in food, particularly in seafood. Exposure to CH3Hg is associated with neurotoxic effects during development. In addition, methylmercury has been classified by the International Agency for Research on Cancer as a possible human carcinogen. Although the diet is known to be the main source of exposure, few studies have characterized the mechanisms involved in the absorption of this contaminant. The present study examines the absorption process using the Caco-2 cell line as a model of the intestinal epithelium. The results indicate that transport across the intestinal cell monolayer in an absorptive direction occurs mainly through passive transcellular diffusion. This mechanism coexists with carrier-mediated transcellular transport, which has an active component. The participation of H(+)- and Na(+)-dependent transport was observed. Inhibition tests point to the possible participation of amino acid transporters (B(0,+) system, L system, and/or y(+)L system) and organic anion transporters (OATs). Our study suggests the participation in CH3Hg absorption of transporters that have already been identified as being responsible for the transport of this species in other systems, although further studies are needed to confirm their participation in intestinal absorption. It should be noted that CH3Hg experiences important cellular acumulation (48-78%). Considering the toxic nature of this contaminant, this fact could affect intestinal epithelium function.


Assuntos
Poluentes Ambientais/farmacologia , Absorção Intestinal , Compostos de Metilmercúrio/farmacologia , Transporte Biológico , Células CACO-2 , Cisteína/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Mucosa Intestinal/metabolismo , Membranas Artificiais , Soroalbumina Bovina/metabolismo , Sódio/farmacologia
19.
J Toxicol Environ Health A ; 77(1-3): 155-68, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24555656

RESUMO

Mercury (Hg) contamination of aquatic systems has been recognized as a global and serious problem affecting both human and environmental health. In the aquatic ecosystems, mercurial compounds are microbiologically transformed with methylation responsible for generation of methylmercury (MeHg) and subsequent biomagnification in food chain, consequently increasing the risk of poisoning for humans and wildlife. High levels of Hg, especially MeHg, are known to exist in Tagus Estuary as a result of past industrial activities. The aim of this study was to isolate and characterize Hg-resistant bacteria from Tagus Estuary. Mercury-resistant (Hg-R) bacteria were isolated from sediments of two hotspots (Barreiro and North Channel) and one reserve area (Alcochete). Mercury contamination in these areas was examined and bacterial susceptibility to Hg compounds evaluated by determination of minimal inhibitory concentrations (MIC). The isolates characterization was based on morphological observation and biochemical testing. Bacteria characteristics, distribution, and Hg resistance levels were compared with metal levels. Barreiro and North Channel were highly contaminated with Hg, containing 126 and 18 µg/g total Hg, respectively, and in Alcochete, contamination was lower at 0.87 µg/g total Hg. Among the isolates there were aerobic and anaerobic bacteria, namely, sulfate-reducing bacteria, and Hg resistance levels ranged from 0.16 to 140 µg/ml for Hg(2+) and from 0.02 to 50.1 µg/ml for MeHg. The distribution of these bacteria and the resistance levels were consistent with Hg contamination along the depth of the sediments. Overall, results show the importance of the characterization of Tagus Estuary bacteria for ecological and human health risk assessment.


Assuntos
Bactérias Aeróbias/efeitos dos fármacos , Bactérias Anaeróbias/efeitos dos fármacos , Estuários , Sedimentos Geológicos/microbiologia , Mercúrio/farmacologia , Poluentes Químicos da Água/farmacologia , Bactérias Aeróbias/isolamento & purificação , Bactérias Anaeróbias/isolamento & purificação , Cromatografia Gasosa , Saúde Ambiental , Monitoramento Ambiental , Sedimentos Geológicos/química , Humanos , Mercúrio/análise , Compostos de Metilmercúrio/análise , Compostos de Metilmercúrio/farmacologia , Testes de Sensibilidade Microbiana , Portugal , Medição de Risco , Espectrofotometria Atômica , Poluentes Químicos da Água/análise , Poluição Química da Água/efeitos adversos , Poluição Química da Água/análise
20.
Pflugers Arch ; 465(12): 1727-40, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23821297

RESUMO

Methylmercury, a potent environmental pollutant responsible for fatal food poisoning, blocked calcium channels of bovine chromaffin cells in a time- and concentration-dependent manner with an IC50 of 0.93 µM. This blockade was not reversed upon wash-out and was greater at more depolarising holding potentials (i.e. 21 % at -110 mV and 60 % at -50 mV, after 3 min perfusion with methylmercury). In ω-toxins-sensitive calcium channels, methylmercury caused a higher blockade of I Ba than in ω-toxins-resistant ones, in which a lower blockade was detected. The sodium current was also blocked by acute application of methylmercury in a time- and concentration-dependent manner with an IC50 of 1.05 µM. The blockade was not reversed upon wash-out of the drug. The drug inhibited sodium current at all test potentials and shows a shift of the I-V curve to the left of about 10 mV. Intracellular dialysis with methylmercury caused no blockade of calcium or sodium channels. Voltage-dependent potassium current was not affected by methylmercury. Calcium- and voltage-dependent potassium current was also drastically depressed. This blockade was related to the prevention of Ca(2+) influx through voltage-dependent calcium channels coupled to BK channels. Under current-clamp conditions, the blockade of ionic current present during the generation and termination of action potentials led to a drastic alteration of cellular excitability. The application of methylmercury greatly reduced the shape and the number of electrically evoked action potentials. Taken together, these results point out that the neurotoxic action evoked by methylmercury may be associated to alteration of cellular excitability by blocking ionic currents responsible for the generation and termination of action potentials.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Compostos de Metilmercúrio/farmacologia , Canais de Sódio/efeitos dos fármacos , Animais , Bário/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Bovinos , Células Cromafins/fisiologia , Concentração Inibidora 50 , Nifedipino/farmacologia , Técnicas de Patch-Clamp , ômega-Agatoxina IVA/farmacologia , ômega-Conotoxina GVIA/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA